1. Introduction
The past two decades have seen a surge in the medical use of nanotechnology. Tens of nanomedicines, mostly liposomes, have been approved by Food and Drug Administration (FDA) or European Medicines Agency (EMA) to treat or diagnose many serious diseases, especially various types of cancer [
1,
2,
3,
4]. Liposomal formulations of doxorubicin (DOX) [
4,
5,
6] represent an important group of nanomedicines, which are indicated for a wide range of cancers. Many studies have demonstrated that, compared with free DOX, DOX-loaded nanomedicines offer substantially lower cardiotoxicity and higher efficacy, both owing to their preferred accumulation at tumor sites [
7,
8].
One abnormal feature in many solid tumors is a fenestrated vasculature [
9], which allows nano-formulations of anticancer drugs to permeate selectively from the blood circulation to the tumor interstitium. The nano-formulations can then accumulate in solid tumors due to their lack of lymphatic drainage, a phenomenon known as the enhanced permeability and retention (EPR) effect. Many long-circulating nano-formulations have been developed to take advantage of the EPR effect, including PEGylated liposomes, hydrophilic polymers, and solid lipid nanoparticles [
10,
11]. However, the fenestrated vasculature distributes mainly in the peripheral of solid tumors, which could limit the distribution of nano-drug formulations to the tumor core, and hence limit their ability to eradicate the entire tumor cell population [
12].
Another abnormality of tumor tissue is its acidic microenvironment. Whereas normal cells in healthy tissues have an intracellular pH (pH
i) of 7.2 and a slightly higher extracellular pH (pH
e) of 7.4, cancer cells in tumors are characterized by a pH
i of 7.2 but a significantly lower pH
e of 6.2–7.0 [
13,
14]. The lower pH
e in the tumor interstitium results from the accumulation of lactate, an acidic by-product of the elevated anaerobic metabolism by the cancer cells in the hypoxic tumor microenvironment [
15]. In response, many pH-sensitive drug delivery systems have been developed, including pH-sensitive liposomes, antibody-drug conjugates with acid-labile linkers, and pH-sensitive polymeric nanoparticles [
16,
17,
18]. A number of pH-sensitive drug delivery systems have shown enhanced anticancer activity compared to their pH-insensitive counterparts in preclinical research [
19,
20]. However, pH-sensitive nano-drug delivery systems have not yet been approved to treat cancer patients.
Imidazole represents an important pH-sensitive functional group in pharmaceutical sciences. Imidazoles carry pKa values around 5.0–6.5 and thus can protonate to assume a positive charge in response to weakly acidic pH in pathophysiological settings [
21]. The incorporation of imidazole-based lipids into nano-formulations has enhanced their intracellular delivery of proteins and nucleic acids [
22,
23]. However, very few studies [
24] have been reported on imidazole-based nano-formulations for anticancer drug delivery.
Herein, we report a novel type of imidazole lipids and their pH-sensitive liposomes. Our goal is to develop imidazole lipids that trigger the liposomes in cooperation with phosphatidylethanolamine-polyethylene glycol conjugates (PE-PEG), which is a key component to stabilize liposomes in blood circulation for anticancer drug delivery. At pH 7.4, the imidazole lipids are mostly uncharged, while at acidic pH, they would protonate and cluster with negatively charged PE-PEG to induce lipid phase conversions. Such liposomes are thus called imidazole-based convertible liposomes (ICL) (
Figure 1). ICL was loaded with the anticancer drug doxorubicin (DOX) and subjected to physicochemical and morphological characterizations. The pH-sensitivity of ICL was assessed by Differential Scanning Calorimetry (DSC), change of ζ-potential, interaction with negatively charged model lipid membranes, and pH-dependent drug release. The anticancer activities of ICL were assessed against both 2D monolayer cancer cells and 3D multicellular tumor spheroids (MCS), which mimic more features of solid tumors than 2D cell cultures, including the acidic microenvironment, the dense ECM, and the hypoxic core [
9,
25]. PEGylated liposomes containing DOX but not the pH-sensitive, imidazole-based lipids were also studied, both as a pH-insensitive control and as a benchmark of clinically used liposomal formulations. The effects of cholesterol on the physicochemical properties, pH-sensitivity and anticancer activities of ICL were also investigated. We report the physicochemical properties and the superior anticancer activities of ICL in both monolayer cancer cells and MCS of multiple cancer cell lines in correlation to their pH-sensitivity.
3. Discussion
Two decades of investigations on nano-drug delivery systems have established the importance of their physicochemical properties in targeting the payload drug to cancer cells (also known as physical targeting) [
37]. Such physicochemical properties include size, shape, surface charge, surface hydrophilicity, drug-loading, and drug release [
37]. The introduction of new characteristics such as active targeting or pH-sensitivity needs to be accomplished in coordination with such properties, which poses a considerable challenge to formulation development.
In this study on ICL, the three imidazole-based lipids triggered PEGylated liposomes by efficiently clustering with phospholipid-PEG conjugates. Such a feature differentiates them from the imidazole lipid reported by Ju et al. [
24] and represents a novel approach to construct stealth liposomes with pH-sensitivity. The clustering action is most probably achieved by the three lipids’ unique structure, in which the imidazole headgroup is linked to the lipid tail at the C2 position through a carbon-sulfur bond so that both nitrogen atoms of the imidazole group can serve as H-bond donors upon protonation at acidic pH (
Figure 2). The protonated imidazole groups can then each bind with negatively charged phosphate groups from two different DPPE-PEG molecules, which in turn crosslink DPPE-PEG molecules into clusters on the ICL surface. As PEGylation serves as a key method to construct long-circulating liposomes for anticancer drug delivery by the EPR effect, the imidazole-based lipids under this study have the potential for wide applications in vivo.
Compared to the doxorubicin-loaded PEG liposomes (Doxil
®) that are in current clinical use, ICL carries the advantage of pH-sensitivity while preserving the physicochemical properties that favor passive targeting to solid tumors. The drug-free ICL carried sizes under 130 nm in diameter while the DOX-loaded ICL formulations carried sizes under or around 200 nm, both of which were within the size range for the EPR effect [
9]. The increase of size and PDI of ICL upon DOX-loading was probably due to the aggregation of DOX molecules with the liposomes because our attempts to load higher concentrations of DOX led to precipitation and because DOX had been reported to aggregate with negatively charged liposomes [
38]. DOX can be loaded into ICL at >50% encapsulation efficiency (EE) and at sufficiently high concentrations (
Table 1) for anticancer studies in cell culture [
39]. The payload DOX concentration of ICL could be further elevated by concentrating DOX-loaded ICL using Tangential Flow Filtration [
40]. Although DOX is elected as the cargo drug in this study for better comparison between ICL and clinically established liposomal formulations, we anticipate that the imidazole lipids under this study can be used to trigger PEGylated liposomes containing various water-soluble anticancer drugs.
In response to the drop of pH, ICL without cholesterol demonstrated a number of substantial changes in their physicochemical properties, including acquisition of positive surface charges (ζ-potential elevation in
Figure 4A), lipid phase separation (DSC studies in
Figure 8A and TEM images in
Figure 7), binding with the bio-mimetic membrane (aggregation with model liposomes in
Figure 5A), and enhanced release of the payload drug DOX (
Figure 6). The extent of most of the changes, namely the positive surface charge acquisition, lipid phase separation, and binding with bio-mimetic membrane, are correlated with higher basicity of the imidazole lipid (DHDMI > DHMI >DHI). This correlation can be explained by our proposed mechanism of ICL’s pH-sensitivity, where the more basic imidazole lipid would be protonated more at the same mildly acidic pH, which would yield more positive charges on liposome surface and more electrostatic interaction between the imidazole lipid and DPPE-PEG, which would, in turn, promote phase separation of ICL membranes and binding between ICL and bio-mimetic membranes. Interestingly, DOX release from ICL did not follow such correlation in that only ICL consisting of DHMI showed substantially enhanced DOX release when the pH dropped from 7.4 to 6.0. TEM images of DHMI/DSPC/DPPE-PEG in comparison to DHI/DSPC/DPPE-PEG and DHDMI/DSPC/DPPE-PEG suggest that this pH-triggered release may be caused by DHMI/DSPC/DPPE-PEG’s unique tendency to collapse into non-lamellar structures at pH 6.0. Alternatively, DHMI/DSPC/DPPE-PEG’s membrane might also have more structural defects at the edge between the separated lipid phases at pH 6.0 to enhance the DOX release.
The incorporation of 25 mol% cholesterol prevented the size increase of ICL during DOX-loading; it also elevated the EE and the payload DOX concentration in ICL. This was probably because cholesterol can improve the stability of the lipid bilayer structure in the ICL formulations. During drug-loading, when the temperature is above the lipid bilayer transition temperature (T > T
m), the liposome membrane was in the fluid phase, in which the lipid molecules were free to move laterally. The addition of cholesterol was found to help suppress the mobility of lipid bilayers in the fluid phase and reduce their permeability to water, thus improving the membrane stability and drug retention during drug loading [
41]. The introduction of cholesterol also diminished the pH-sensitivity of ICL (
Figure 4,
Figure 5,
Figure 6 and
Figure 7), probably because the incorporation of cholesterol obstructed the lateral movements of lipids in bilayers at T < T
m. The nonpolar cholesterol molecules were found to tie up the neighboring lipid’s hydrocarbon chains to minimize cholesterol molecules’ thermodynamically unfavorable exposure to water at the membrane-water interface [
42]. In ICL, such cholesterol-lipid interaction would substantially limit the movement of the imidazole-based lipids and DPPE-PEG, thus hindering their clustering at acidic pH. Furthermore, ICL with cholesterol maintained negative ζ-potentials at acidic pH, indicating that the protonation of the imidazole-based lipids was also suppressed by cholesterol (
Figure 4B). This is probably because the addition of cholesterol increases the hydrophobicity of the liposome membrane, which in turn reduces its affinity with cations [
43].
The targeting of cytotoxic chemotherapy drugs to enhance their efficacy and safety is a complicated process with multiple challenges that all need to be addressed by the drug delivery system, including preferred distribution of the drug molecules from blood circulation to the tumor interstitium, sufficient permeation of the drug molecules to all areas of the solid tumor, and uptake of the drug molecules by virtually all the cancer cells in the tumor. It is therefore critical that anticancer drug delivery systems are evaluated by biological models that simulate these multiple challenges. ICL formulations under this study were evaluated by both monolayer cancer cells and 3D MCS in culture in order to test the potential of their pH-sensitivity to enhance the anticancer activity [
34].
In monolayer Hela cells, ICL’s higher ability to suppress cell viability is strongly correlated with lower pH and higher pKa, which are both correlated with the acquisition of positive charges on the ICL surface, ICL’s phase separation, and ICL’s interaction with negatively charged bio-mimetic membranes. Such strong correlations suggest that ICL’s activities to suppress monolayer cell viabilities can be attributed to ICL’s enhanced binding to the cancer cells at lowered pH. More specifically, the drop of pH protonates the imidazole lipids, which cluster DPPE-PEG lipids to expose a de-PEGylated and positively charged ICL surface, which in turn binds to the cancer cell surface to induce the endocytosis of the DOX-loaded ICL and consequently the cell death.
The ICL’s pattern of suppressing the cell viability in 3D multicellular spheroids was quite different from that in 2D monolayer cells. Overall, ICL consisting of DHMI, the imidazole lipid of the second-highest calculated pKa (6.20 ± 0.5), yielded the highest activity to suppress MCS viability, rather than DHDMI of the highest calculated pKa (6.75 ± 0.5). This is probably due to the dynamic balance between the binding of ICL to the cancer cells in MCS and the penetration of ICL to reach the most cancer cells in MCS. On the one hand, DHI may carry too low a pKa (5.53 ± 0.5) to sufficiently trigger its ICL in the mildly acidic microenvironment of MCS; on the other hand, DHDMI may carry too high a pKa, which would trigger most of its ICL to bind to only the cancer cells in the peripheral region of MCS. Paradoxically, DHMI may carry the optimal basicity (pKa 6.20 ± 0.5, closest to the measured interstitial pH of MCS) to facilitate both the penetration and the cellular binding of its ICL. Furthermore, among all the ICL under this study, DHMI/DSPC/DPPE-PEG showed the unique property of pH-enhanced drug release, which would allow such ICL to selectively release DOX in the MCS interstitium to kill multiple adjacent cancer cells, also known as the bystander effect [
44].
4. Materials and Methods
4.1. Materials
1,2-Di-O-hexadecyl-rac-glycerol (DHG), 2-mercaptoimidazole, 4-methyl-1H-imidazole-2-thiol, and 4,5-dimethyl-1H-imidazole-2-thiol were purchased from Santa Cruz Biotechnology (Dallas, TX, USA). p-Toluenesulfonyl chloride, 2-[4-(2-hydroxyethyl)piperazin-1-yl]-ethanesulfonic acid (HEPES), and 2-(N-morpholino)ethanesulfonic acid (MES) were purchased from Fisher Scientific (Hampton, NH, USA). Triethylamine (TEA) was purchased from Alfa Aesar (Haverhill, MA, USA). The lipids 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-[azido(polyethylene glycol)-2000 (DPPE-PEG (2000)), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE), 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-L-serine (sodium salt) (POPS) and L-α-phosphatidylinositol (Soy) (L-R-PI) were purchased from Avanti Polar Lipids, Inc. (Alabaster, AL, USA). Cholesterol, Dowex® 50WX-4 (50–100 mesh), Sephadex G-25, and Uranyl acetate (UA) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Doxorubicin hydrochloride was purchased from Biotang (Waltham, MA, USA). Carbon-coated copper grids (200 mesh) for electron microscopy were purchased from Polysciences (Warrington, PA, USA). The HeLa, A549, MDA-MB-231 and MDA-MB-468 cell lines were purchased from ATCC (Manassas, VA, USA). The Dulbecco’s Modified Eagle’s Medium (DMEM), Advanced DMEM/F12 medium, Trypsin-EDTA, L-glutamine, fetal bovine serum, and collagen were purchased from Thermo-Fisher Scientific (Waltham, MA, USA). The RPMI 1640 medium, penicillin-streptomycin, 96-well Ultra-low Attachment round-button microplates, 96-well solid white microplates, CellTiter-Glo 3D cell viability assay kits and MTS CellTiter 96® AQueous One Solution cell proliferation assay kits (Promega Corp., WI, USA) were purchased from VWR (Radnor, PA, USA). All other organic solvents and chemicals were purchased from Sigma-Aldrich (St. Louis, MO, USA), Fisher Scientific (Hampton, NH, USA) or VWR (Radnor, PA, USA).
4.2. Synthesis of 2,3-di-O-Hexadecyl-1-rac-glyceryl-tosylate (DHG-Tosylate)
1,2-Di-O-hexadecyl-rac-glycerol (DHG) (2.30 g, 4.25 mmol) was mixed with anhydrous dichloromethane (20 mL) and pyridine (18.6 mL, 225 mmol). p-Toluenesulfonyl chloride (1.90 g, 9.97 mmol) was dissolved in ~0.5 mL anhydrous dichloromethane and transferred into the mixture. The reaction mixture was stirred under argon at room temperature for 8 to 12 h. The reaction mixture was then mixed well with 10 mL anhydrous dichloromethane and washed with saturated Na2CO2 solution 3 times. The organic phase was separated from the aqueous phase, dried with MgSO4, filtered, and then evaporated into dryness under vacuum. The resultant residue was separated by silica gel chromatography with dichloromethane as the mobile phase to yield 2.53 g solid (86%). DART Mass Spectrum: 695.5; calculated, 695.6 (MH)+. 1H-NMR (600 MHz, CDCl3, δ ppm): 0.87 (t, 6H, 2 CH3(CH2)15-), 1.18–1.31 (m, 52H, 2 OCH2CH2(CH2)13CH3), 1.46 (m, 4H, 2 OCH2CH2(CH2)13CH3), 2.44 (s, 3H, -(C6H4)CH3), 3.31–3.62 (m, 7H, glyceryl/hexadecyl -CH2O and -CHO-), δ 4.14 (m, 2H, -CH2OSO2-), δ 7.33 (d, 2H, aromatic protons ortho to -CH3), and δ 7.78 (d, 2H, aromatic protons ortho to -SO2-).
4.3. Synthesis of sn-2-((2,3-Dihexadecyloxypropyl)thio)-1H-imidazole (DHI)
2-Mercaptoimidazole (0.91 g, 9.06 mmol) was dissolved in 8–9 mL of anhydrous N, N-dimethylformamide (DMF). DHG-tosylate (1.265 g, 1.82 mmol) was dissolved in 7–8 mL of anhydrous dichloromethane and transferred into the above-mentioned solution, followed by the addition of triethylamine (TEA, 1.27 mL, 9.08 mmol). The reaction mixture was stirred under argon at 55 °C for 48 h. The solvent was evaporated under a vacuum, and the resultant residue was dissolved in dichloromethane. The solution was washed with saturated sodium bicarbonate solution 3 times, dried with sodium carbonate, filtered, and then evaporated into dryness under vacuum. The resultant residue was then separated by silica gel chromatography with 1–5 vol% methanol in dichloromethane as the mobile gradient phase to yield DHI (25–30%). DART Mass Spectrum: 623.48 (
Figure S1); calculated, 623.55 (MH)
+. 1H-NMR (
Figure S2, 600 MHz, CDCl
3): δ 0.87 (t, 6H, 2 C
H3(CH
2)
15-), δ 1.19–1.32 (m, 54H, 2 -OCH
2CH
2(C
H2)
13CH
3 and -
H2CSCNH-), δ 1.55 (m, 4H, 2 OCH
2C
H2(CH
2)
13CH
3, δ 3.2–3.7 (m, 7H, glyceryl/hexadecyl -C
H2O and -C
HO-), δ 7.02 (d, 1H, H
2CSC-NHC
H=CH-N=), δ 7.21 (d, 1H, H
2CSC-NHCH=C
H-N=). Elemental analysis: C 73.27%, H 12.25%, N 4.56%; calculated: C 73.25%, H 11.97%, N 4.50%. Calculated pK
a using ACD/pKa DB software: 5.53 ± 0.5.
4.4. Synthesis of sn-2-((2,3-Dihexadecyloxypropyl)thio)-5-methyl-1H-imidazole (DHMI)
4-Methyl-1H-imidazole-2-thiol (1.03 g, 9.03 mmol) was used to prepare DHMI, using the same synthesis method as DHI. Yield: 25–30%. DART Mass Spectrum: 637.55 (
Figure S1); calculated, 637.57 (MH)
+. 1H-NMR (
Figure S2, 600 MHz, CDCl
3): δ 0.87 (t, 6H, 2 C
H3(CH
2)
15-), δ 1.19–1.34 (m, 54H, 2 -OCH
2CH
2(C
H2)
13CH
3 and -
H2CSCNH-), δ 1.53 (m, 4H, 2 OCH
2C
H2(CH
2)
13CH
3), δ 2.41 (s, 3H, -H
2CSC-NH-C(C
H3)-), δ 3.2–3.7 (m, 7H, glyceryl/hexadecyl -C
H2O and -C
HO-), δ 6.81 (s, 1H, -H
2CSC=N-C
H=). Elemental analysis: C 73.63%, H 12.08%, N 4.35%; calculated: C 73.52%, H 12.02%, N 4.40%. Calculated pK
a using ACD/pKa DB software: 6.20 ± 0.5.
4.5. Synthesis of sn-2-((2,3-Dihexadecyloxypropyl)thio)-4,5-dimethyl-1H-imidazole (DHDMI)
4,5-Dimethyl-1H-imidazole-2-thiol (1.15 g, 9.03 mmol) was used to prepare DHDMI, using the same synthesis method as DHI. Yield: 25–30%. DART Mass Spectrum: 651.56 (
Figure S1); calculated, 651.59 (MH)
+. 1H-NMR (
Figure S2, 600 MHz, CDCl
3): δ 0.87 (t, 6H, 2 C
H3(CH
2)
15–), δ 1.19–1.34 (m, 54H, 2 -OCH
2CH
2(C
H2)
13CH
3 and -
H2CSCNH-), δ 1.53 (m, 4H, 2 OCH
2C
H2(CH
2)
13CH
3), δ2.22 (s, 3H, -H
2CSC-NHC(C
H3)=), δ2.24 (s, 3H, -H
2CSC=N-C(C
H3)=), δ 3.3–3.7 (m, 7H, glyceryl/hexadecyl -C
H2O and -C
HO-). Elemental analysis: C 73.78%, H 12.24%, N 4.16%; calculated: C 73.78%, H 12.07%, N 4.30%. Calculated pK
a using ACD/pKa DB software: 6.72 ± 0.5.
4.6. Preparation of ICL Formulations
A dichloromethane solution of an imidazole-based lipid and a chloroform solution of other lipids were mixed in a round-bottom flask. The organic solvents were evaporated under reduced pressure to form a lipidic film at 70 °C. The lipidic film was further dried in a high vacuum for over 4 h at room temperature to remove the residual solvent. The lipidic film was then hydrated with HEPES buffer (pH 7.4, 30 mM HEPES) containing 300 mM MnSO
4 by intermittent agitation in a 70 °C water bath under argon to obtain a liposome suspension containing 20 mM total lipids. The liposome suspension was freeze-anneal-thawed by rapidly freezing in liquid nitrogen, immerging in the ice-water mixture for 2 min and incubating in a 70 °C water bath for 4 min. The freeze-anneal-thawing was repeated 11 times. The liposome suspension was sequentially extruded 21 times each through 400 nm, 200 nm and 100 nm polycarbonate membranes (Nucleopore Corp., Pleasanton, CA, USA) using a hand-held Mini-extruder (Avanti Polar Lipids Inc., Alabaster, AL, USA) at 70 °C to reduce and homogenize the size of liposomes. DOX was then loaded into the liposomes as follows, using a transmembrane MnSO
4 gradient [
45]. The extruded liposomes were separated from the unencapsulated MnSO4 by size exclusion chromatography using a Sephadex G-75 column pre-equilibrated with isotonic HEPES buffer (pH 7.4, 5 mM HEPES, 140 mM NaCl). DOX (0.75 mg/mL) dissolved in the same isotonic HEPES buffer was then mixed with the liposome suspension in a 1:2 (
v/
v) ratio, and the mixture was incubated in a 70 °C water bath for 90 min. The cation-exchange resin Dowex
® 50WX-4, 50–100 mesh was pre-treated with NaOH and NaCl [
46], mixed with the DOX-liposome mixture at DOX: resin = 1:60 (
w/
w), and then shaken gently for 25 min to remove the unencapsulated DOX from the DOX-loaded liposomes. The resin was separated from the DOX-loaded liposomes by filtration. A tangential flow filtration column (MicroKros
®, Spectrum, Stamford, CT, USA) was used to concentrate the liposome suspension by partially removing the extra-liposomal buffer. Typically, a 2 mL liposome suspension was extruded 14 times through the tangential flow filtration column to yield a ~0.5 mL concentrated formulation. The lipid composition of the liposomes under study is listed in
Table 3.
4.7. Size Measurement
An aliquot (2.5–5 μL) of a liposome suspension was diluted in 150 μL isotonic buffer, and the size was measured at room temperature by dynamic light scattering (Zetasizer ZS90, Malvern Instruments Ltd., Malvern, Malvern, UK). The data were analyzed based on light intensity distribution to give hydrodynamic diameters.
4.8. Quantification of Encapsulation Efficiency (EE)
An aliquot (10 μL) of DOX-loaded liposome suspension was lysed with 90 μL lysis buffer (90% (
v/
v) isopropanol, 0.075 M HCl) [
46] in a 96-well Black Clear Bottom Polystyrene microplate (Corning
®, NY, USA), together with 10 μL DOX standard solutions diluted in the same lysing buffer (90 μL). The microplate was covered with foil, and the fluorescence of the samples (λ
ex = 486 nm, λ
em = 590 nm) was recorded on a Synergy HT microplate reader (Biotek, Winooski, VT, USA). The concentration of the payload DOX of liposomes was estimated using a standard calibration curve from the fluorescence of the DOX standard solutions. The encapsulation efficiency (EE) of the liposomes was then calculated by the following formula.
4.9. Differential Scanning Calorimetry
A VP-DSC Instrument (MicroCal, LLC, Northampton, MA, USA) was used for the differential scanning calorimetry (DSC) studies. DSC scans were performed on 0.5 mL liposome suspensions containing 2.5 mM total lipids at pH 7.4 and pH 6.0. The thermograms of liposome suspensions were acquired from 40 °C to 75 °C at a scan rate of 5 °C/h. Each excess heat capacity curve of a liposome sample was normalized by subtraction of the thermogram of the buffer acquired simultaneously under identical conditions.
4.10. pH-Triggered Change of ζ-Potential
In order to enhance the detection of changes in liposome surface charge, the liposomes were prepared by hydration in an isotonic buffer of low ionic strength (pH 7.4, 5 mM HEPES, 5% (
w/
v) Glucose) [
47]. Aliquots (50–100 μL) of the resultant liposome suspensions were diluted in 900 μL isotonic MES buffer (final pH 6.0 and 6.5, 10 mM MES, 5% (
w/
v) Glucose) and 900 μL isotonic HEPES buffer (final pH 7.0 and 7.4, 10 mM HEPES, 5% (
w/
v) Glucose). The
ζ-potential was then measured at 37 °C based on electrophoresis mobility under applied voltage (Zetasizer ZS90, Malvern Instruments Ltd., Malvern, UK).
4.11. pH-Dependent Interaction with Model Liposomes
The model liposomes (POPC:POPE:POPS:L-R-PI:cholesterol = 50:20:5:10:15 (mol%)) mimicking the lipid composition and surface charge of biomembranes were prepared based on a previous report [
31]. As measured by Zetasizer ZS90, the mean size of the model liposomes was 192.7 nm in diameter, and the
ζ-potential was −51.77 ± 1.18 mV. Suspensions of ICL and pH-insensitive control liposomes were each mixed with the model liposomes at a 1:1 total lipid molar ratio. An aliquot (5 μL) of each mixture was diluted in 150 μL isotonic MES buffer (final pH 6.0 and 6.5, 10 mM MES, 140 mM NaCl) and isotonic HEPES buffer (final pH 7.0 and 7.4, 10 mM HEPES, 140 mM NaCl), and incubated at 37 °C for 5 min. The particle size of the diluted mixtures was measured at 37 °C by dynamic light scattering (Zetasizer ZS90, Malvern Instruments Ltd., UK).
4.12. pH-Dependent Drug Release
Each liposome formulation was severally diluted (100 μL aliquots) with 500 μL MES buffer (final pH 6.0 and 6.5, 100 mM MES, 1.7% (
w/
v) Glucose) and HEPES buffer (final pH 7.0 and 7.4, 100 mM HEPES, 1.7% (
w/
v) Glucose). An aliquot (10 μL) of each diluted liposome formulation was immediately lysed with 90 μL lysis buffer (90% (
v/
v) isopropanol, 0.075 M HCl) in a 96-well Black Clear Bottom Polystyrene microplate and the initial DOX concentration
Ci (as at the 0-h time point) was qualified with standard DOX solutions. The liposome samples diluted by buffers at various pH (6.0, 6.5, 7.0, 7.4) were then mixed with cation-exchange resin Dowex
® 50WX-4 (50–100 mesh) at DOX:resin = 1:200 (
w/
w) ratio. The mixtures were incubated and gently shaken at 37 °C. At different time points (1, 3, 6, 12 h), each mixture was allowed to settle briefly, and an aliquot (10 μL) of the resultant supernatant was harvested, lysed, and its DOX concentration measured as mentioned before. The percentage of DOX release was determined by the following equation,
where C
s is the concentration of DOX in the supernatant of the liposome-resin mixture at different time points, C
i is the initial liposomal DOX concentration.
4.13. Transmission Electron Microscopy
The morphology of ICL formulations was observed on a JEOL-JEM 1230 Electron Microscope (JEOL, Tokyo, Japan). Carbon-coated copper TEM grids (200 mesh) were subjected to glow discharge before usage to increase their hydrophilicity. An aliquot (5 µL) of diluted ICL suspension (approximately 1 mM total lipids) at pH 7.4 or 6.0 was dripped onto the grid to wet its surface for 1 min and then blotted with filter paper to generate a thin film. The sample film was then wetted five times with 5 µL of the negative stain 2% uranyl acetate (UA) between blotting. The grid was dried at room temperature and then transferred into the electron microscope for imaging at an accelerating voltage of 100 kV. The samples of ICL mixed with model liposomes were prepared and imaged by the same method.
4.14. Cell Culture
Cervical cancer cell line HeLa, lung cancer cell line A549, and breast cancer cell lines MDA-MB-231 and MDA-MB-468 were cultured to construct 3D MCS in order to evaluate the anticancer activities of ICL. Hela cells were also cultured into monolayer cells. HeLa cells were maintained in DMEM media; A549 cells were maintained in RPMI 1640 media; MDA-MB-231 and MDA-MB-468 cells were maintained in advanced DMEM/F12 media. All media were supplemented with 10% fetal bovine serum, 1% Penicillin-Streptomycin, and 1% L-glutamine. All cells were grown in a humidified atmosphere of 5% CO2 in air at 37 °C and passaged at 85% confluence. In all studies, the cells were sub-cultured every 2–3 days and used for experiments at passages 5–20.
4.15. Cytotoxicity Assays on 2D Monolayer Hela Cells
Monolayer HeLa cells at ~85% confluence were suspended by trypsinization, and the cell density was determined with a Handheld Automated Cell Counter (Millipore, Burlington, MA, USA). The cells were then diluted to ~80,000 cells/mL in complete growth media and seeded into 96-well Clear Microplates (Corning, NY, USA) at ~8000 cells/well by transferring 100 μL of the cell suspension into each well. The cytotoxicity assay was carried out on the cells 8 h after they were seeded. The cells were washed with PBS and treated with DOX-loaded liposomes or free DOX solutions in complete growth media at incremental concentrations. The pH of the media (10 mL) was adjusted to 7.4, 7.0, 6.5, and 6.0 with glacial acetic acid. After 12-h incubation, the media was removed, and the cells were washed with 100 μL PBS buffer and supplemented with 100 μL/well complete growth media and 20 µL/well MTS CellTiter 96® AQueous One Solution. The mixture was incubated for 4 h at 37 °C with 5% CO2. The cell viability was quantified by UV/visible absorbance at 490 nm on a Synergy HT microplate reader (Biotek, Winooski, VT, USA). The Hela cells treated with growth media at corresponding pHs without free DOX or DOX-loaded liposomes were referred to as 100% cell viability.
4.16. Cytotoxicity Assays on 3D Multicellular Spheroids
Monolayer cells in T75 flasks were trypsinized, and the cell density in the suspensions was determined with a Handheld Automated Cell Counter. The cells were then seeded into 96-well Ultra-low Attachment (ULA) round-bottom microplates (Corning, NY, USA) at ~500 Hela cells/well, ~5000 A549 cells/well, ~3000 MDA-MB-231 cells/well, and ~2000 MDA-MB-468 cells/well by transferring 100 μL properly diluted cell suspensions in complete growth media containing collagen (0% for HeLa, 0.3% for A549, 1% for MDA-MB-231, and 1% for MDA-MB-468 cell lines). If needed, the microplates were centrifuged at 7 °C to promote cell aggregation (
Table S1). Complete growth media (100 μL) was added to each well on the second day after seeding. The growth media were then partially exchanged every other day by replacing 100 μL of media in each well with 100 μL fresh media to maintain a 200 μL/well total media volume. The cytotoxicity assays were carried out on selected MCS whose diameter reached or exceeded 500 μm (
Figure S3) [
48,
49]. Part of the growth media (100 μL/well) was replaced with the same volume of DOX-loaded liposomes or free DOX solutions in complete growth media at incremental concentrations. After 72 h incubation, each MCS was transferred into a well of a 96-well Solid White microplate (Corning, NY, USA) together with 100 μL media. Then, 100 μL reagent of the CellTiter-Glo 3D cell viability assay was then added to each well, and the microplate was covered with foil, shaken on an orbital shaker for 5 min, and then incubated for 25 min at room temperature. The viability of MCS was then measured by luminescence intensity on a Synergy HT microplate reader (Biotek, Winooski, VT, USA). The MCS treated by growth media without free DOX or DOX-loaded liposomes was referred to as 100% cell viability.